1
|
O'Brien EA, Purslow JA, Wall BJ, VanVeller B. Hydrogen-bonding behavior of amidines in helical structure. Chem Sci 2024:d4sc06108j. [PMID: 39479155 PMCID: PMC11514129 DOI: 10.1039/d4sc06108j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Amidines are an isostere of the amide bond and are completely unexplored in peptide secondary structure. This study marks the first investigation of the structural implications of amidines in folded helices. Amidines were found to engage in hydrogen-bonding interactions that are compatible with helical structure. The protic state of the amidine is also adaptive to local interactions, able to form stronger hydrogen bonds with proton donors or form the first example of a salt bridge along the peptide backbone to stabilize the C-terminus of the helical fold. The rationalization of this behavior was aided by our discovery that the basicity of amidines within peptide backbones can be significantly lower than previously assumed for small molecules. These findings compel investigation of amidines in peptide-drug design.
Collapse
Affiliation(s)
- Emily A O'Brien
- Department of Chemistry, Iowa State University Ames IA 50011 USA
| | | | - Brendan J Wall
- Department of Chemistry, Iowa State University Ames IA 50011 USA
| | - Brett VanVeller
- Department of Chemistry, Iowa State University Ames IA 50011 USA
| |
Collapse
|
2
|
Mauran L, Assailly C, Goudreau SR, Odaert B, Guichard G, Pasco M. Short Oligourea Foldamers as N- or C-Caps for Promoting α-Helix Formation in Water. Chembiochem 2024; 25:e202400427. [PMID: 38943628 DOI: 10.1002/cbic.202400427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024]
Abstract
While foldamers have been extensively studied as protein mimics and especially as α-helix mimics, their use as capping motif to enhance α-helix propensity remains comparatively much limited. In this study, we leverage the structural similarities between urea-based helical foldamers and α-helix to investigate the efficacy of oligoureas as N- or C-caps for reinforcing α-helical structures in water. Short oligoureas, comprising 3 to 4 residues, were strategically introduced at the N- or C-terminus of two peptide sequences (S-peptide and an Ala-rich model sequence). The impact of these foldamer insertions on peptide conformation was examined using electronic circular dichroism (ECD) and solution NMR. This research identifies specific foldamer sequences capable of promoting α-helicity when incorporated at either terminus of the peptides. Not only does this work broaden the application scope of foldamers, but it also provides valuable insights into novel strategies for modulating peptide conformation in aqueous environments. The findings presented in this study may have implications for peptide design and the development of bioactive foldamer-based peptide mimics.
Collapse
Affiliation(s)
- Laura Mauran
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
- IMMUPHARMA BIOTECH SAS, 15 rue de Bruxelles, 75009, Paris, France
| | - Coralie Assailly
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | | | - Benoît Odaert
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600, Pessac, France
| | - Gilles Guichard
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Morgane Pasco
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| |
Collapse
|
3
|
Naskar S, Minoia A, Duez Q, Izuagbe A, De Winter J, Blanksby SJ, Barner-Kowollik C, Cornil J, Gerbaux P. Polystyrene Chain Geometry Probed by Ion Mobility Mass Spectrometry and Molecular Dynamics Simulations. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2408-2419. [PMID: 39279164 DOI: 10.1021/jasms.4c00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Polystyrene (PS) is a thermoplastic polymer commonly used in various applications due to its bulk properties. Designing functional polystyrenes with well-defined structures for targeted applications is of significant interest due to the rigid and apolar nature of the polymer chain. Progress is hindered to date by the limitations of current analytical methods in defining the atomistic-level folding of the polymer chain. The integration of ion mobility spectrometry and molecular dynamics simulations is beneficial in addressing these challenges. However, data on gas-phase polystyrene ions are rarely reported in the literature. We herein investigate the gas phase structure of polystyrene ions with different end groups to establish how the nature and the rigidity of the monomer unit affect the charge stabilization. We find that, in contrast to polar polymers in which the charges are located deep in the ionic globules, the charges in the PS ions are rather located at the periphery of the polymer backbone, leading to singly and doubly charged PS ions adopting dense elliptic-shaped structures. Molecular dynamics (MD) simulations indicate that the folding of the PS rigid chain is controlled by phenyl ring interactions with the charge ultimately remaining excluded from the core of the globular ions, whereas the folding of polyether ions is initiated by the folding of the flexible polyether chain around the sodium ion that remains deeply enclosed in the core of the ions.
Collapse
Affiliation(s)
- Sarajit Naskar
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
- Center for Materials Science, School of Chemistry and Physics, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| | - Andrea Minoia
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| | - Quentin Duez
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| | - Aidan Izuagbe
- Center for Materials Science, School of Chemistry and Physics, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
| | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| | - Stephen J Blanksby
- Center for Materials Science, School of Chemistry and Physics, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
| | - Christopher Barner-Kowollik
- Center for Materials Science, School of Chemistry and Physics, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
| | - Jérôme Cornil
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| | - Pascal Gerbaux
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons - UMONS, 23 Place du Parc, B-7000 Mons, Belgium
| |
Collapse
|
4
|
Hink F, Aduriz-Arrizabalaga J, Lopez X, Suga H, De Sancho D, Rogers JM. Mixed Stereochemistry Macrocycle Acts as a Helix-Stabilizing Peptide N-Cap. J Am Chem Soc 2024; 146:24348-24357. [PMID: 39182188 DOI: 10.1021/jacs.4c05378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interactions between proteins and α-helical peptides have been the focus of drug discovery campaigns. However, the large interfaces formed between multiple turns of an α-helix and a binding protein represent a significant challenge to inhibitor discovery. Modified peptides featuring helix-stabilizing macrocycles have shown promise as inhibitors of these interactions. Here, we tested the ability of N-terminal to side-chain thioether-cyclized peptides to inhibit the α-helix binding protein Mcl-1, by screening a trillion-scale library. The enriched peptides were lariats featuring a small, four-amino-acid N-terminal macrocycle followed by a short linear sequence that resembled the natural α-helical Mcl-1 ligands. These "Heliats" (helical lariats) bound Mcl-1 with tens of nM affinity, and inhibited the interaction between Mcl-1 and a natural peptide ligand. Macrocyclization was found to stabilize α-helical structures and significantly contribute to affinity and potency. Yet, the 2nd and 3rd positions within the macrocycle were permissible to sequence variation, so that a minimal macrocyclic motif, of an N-acetylated d-phenylalanine at the 1st position thioether connected to a cysteine at the 4th, could be grafted into a range of peptides and stabilize helical conformations. We found that d-stereochemistry is more helix-stabilizing than l- at the 1st position in the motif, as the d-amino acid can utilize polyproline II torsional angles that allow for more optimal intrachain hydrogen bonding. This mixed stereochemistry macrocyclic N-cap is synthetically accessible, requiring only minor modifications to standard solid-phase peptide synthesis, and its compatibility with peptide screening can provide ready access to helix-focused peptide libraries for de novo inhibitor discovery.
Collapse
Affiliation(s)
- Fabian Hink
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Julen Aduriz-Arrizabalaga
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Xabier Lopez
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Bunkyo-ku 113-0033, Japan
| | - David De Sancho
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Joseph M Rogers
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
5
|
DiGiorno MC, Vithanage N, Victorio CG, Kreitler DF, Outlaw VK, Sawyer N. Structural Characterization of Disulfide-Linked p53-Derived Peptide Dimers. RESEARCH SQUARE 2024:rs.3.rs-4644285. [PMID: 39070635 PMCID: PMC11275974 DOI: 10.21203/rs.3.rs-4644285/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Disulfide bonds provide a convenient method for chemoselective alteration of peptide and protein structure and function. We previously reported that mild oxidation of a p53-derived bisthiol peptide (CTFANLWRLLAQNC) under dilute non-denaturing conditions led to unexpected disulfide-linked dimers as the exclusive product. The dimers were antiparallel, significantly α-helical, resistant to protease degradation, and easily reduced back to the original bisthiol peptide. Here we examine the intrinsic factors influencing peptide dimerization using a combination of amino acid substitution, circular dichroism (CD) spectroscopy, and X-ray crystallography. CD analysis of peptide variants suggests critical roles for Leu6 and Leu10 in the formation of stable disulfide-linked dimers. The 1.0 Å resolution crystal structure of the peptide dimer supports these data, revealing a leucine-rich LxxLL dimer interface with canonical knobs-into-holes packing. Two levels of higher-order oligomerization are also observed in the crystal: an antiparallel "dimer of dimers" mediated by Phe3 and Trp7 residues in the asymmetric unit and a tetramer of dimers mediated by Trp7 and Leu10. In CD spectra of Trp-containing peptide variants, minima at 227 nm provide evidence for the dimer of dimers in dilute aqueous solution. Importantly, and in contrast to the original dimer model, the canonical leucine-rich core and robust dimerization of most peptide variants suggests a tunable molecular architecture to target various proteins and evaluate how folding and oligomerization impact various properties, such as cell permeability.
Collapse
|
6
|
Qin W, Wei X, Yang D, Luo Q, Huang M, Xing S, Wei W, Liang L, Huang J, Zhou Z, Lu F. Ras-Targeting Stabilized Peptide Increases Radiation Sensitivity of Cancer Cells. Bioconjug Chem 2024; 35:737-743. [PMID: 38738511 DOI: 10.1021/acs.bioconjchem.4c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Radiation therapy is one of the most common treatments for cancer. However, enhancing tumors' radiation sensitivity and overcoming tolerance remain a challenge. Previous studies have shown that the Ras signaling pathway directly influences tumor radiation sensitivity. Herein, we designed a series of Ras-targeting stabilized peptides, with satisfactory binding affinity (KD = 0.13 μM with HRas) and good cellular uptake. Peptide H5 inhibited downstream phosphorylation of ERK and increased radio-sensitivity in HeLa cells, resulting in significantly reduced clonogenic survival. The stabilized peptides, designed with an N-terminal nucleation strategy, acted as potential radio-sensitizers and broadened the applications of this kind of molecule. This is the first report of using stabilized peptides as radio-sensitizers, broadening the applications of this kind of molecule.
Collapse
Affiliation(s)
- Weirong Qin
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Xiangzan Wei
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biological Molecular Medicine Research (Guangxi Medical University), Nanning 530021, Guangxi, P. R. China
| | - Dan Yang
- Department of Science & Technology of Shandong Province, Jinan 250101, Shandong, P. R. China
| | - Qinhong Luo
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, P. R. China
| | - Mingyu Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Shangping Xing
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Wei Wei
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Lin Liang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Jin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China
| | - Ziyuan Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, P. R. China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong, P. R. China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, P. R. China
| |
Collapse
|
7
|
van der Walt M, Möller DS, van Wyk RJ, Ferguson PM, Hind CK, Clifford M, Do Carmo Silva P, Sutton JM, Mason AJ, Bester MJ, Gaspar ARM. QSAR Reveals Decreased Lipophilicity of Polar Residues Determines the Selectivity of Antimicrobial Peptide Activity. ACS OMEGA 2024; 9:26030-26049. [PMID: 38911757 PMCID: PMC11191095 DOI: 10.1021/acsomega.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024]
Abstract
Antimicrobial resistance has increased rapidly, causing daunting morbidity and mortality rates worldwide. Antimicrobial peptides (AMPs) have emerged as promising alternatives to traditional antibiotics due to their broad range of targets and low tendency to elicit resistance. However, potent antimicrobial activity is often accompanied by excessive cytotoxicity toward host cells, leading to a halt in AMP therapeutic development. Here, we present multivariate analyses that correlate 28 peptide properties to the activity and toxicity of 46 diverse African-derived AMPs and identify the negative lipophilicity of polar residues as an essential physiochemical property for selective antimicrobial activity. Twenty-seven active AMPs are identified, of which the majority are of scorpion or frog origin. Of these, thirteen are novel with no previously reported activities. Principal component analysis and quantitative structure-activity relationships (QSAR) reveal that overall hydrophobicity, lipophilicity, and residue side chain surface area affect the antimicrobial and cytotoxic activity of an AMP. This has been well documented previously, but the present QSAR analysis additionally reveals that a decrease in the lipophilicity, contributed by those amino acids classified as polar, confers selectivity for a peptide to pathogen over mammalian cells. Furthermore, an increase in overall peptide charge aids selectivity toward Gram-negative bacteria and fungi, while selectivity toward Gram-positive bacteria is obtained through an increased number of small lipophilic residues. Finally, a conservative increase in peptide size in terms of sequence length and molecular weight also contributes to improved activity without affecting toxicity. Our findings suggest a novel approach for the rational design or modification of existing AMPs to increase pathogen selectivity and enhance therapeutic potential.
Collapse
Affiliation(s)
- Mandelie van der Walt
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Dalton S. Möller
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Rosalind J. van Wyk
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Philip M. Ferguson
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Charlotte K. Hind
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - Melanie Clifford
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - Phoebe Do Carmo Silva
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - J. Mark Sutton
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - A. James Mason
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Megan J. Bester
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Anabella R. M. Gaspar
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
8
|
Xue J, Fu Y, Li H, Zhang T, Cong W, Hu H, Lu Z, Yan F, Li Y. All-hydrocarbon stapling enables improvement of antimicrobial activity and proteolytic stability of peptide Figainin 2. J Pept Sci 2024; 30:e3566. [PMID: 38271799 DOI: 10.1002/psc.3566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024]
Abstract
Figainin 2 is a cationic, hydrophobic, α-helical host-defense peptide with 28 residues, which was isolated from the skin secretions of the Chaco tree frog. It shows potent inhibitory activity against both Gram-negative and Gram-positive pathogens and has garnered considerable interest in developing novel classes of natural antibacterial agents. However, as a linear peptide, conformational flexibility and poor proteolytic stability hindered its development as antibacterial agent. To alleviate its susceptibility to proteolytic degradation and improve its antibacterial activity, a series of hydrocarbon-stable analogs of Figainin 2 were synthesized and evaluated for their secondary structure, protease stability, antimicrobial, and hemolytic activities. Among them, F2-12 showed significant improvement in protease resistance and antimicrobial activity compared to that of the template peptide. This study provides a promising strategy for the development of antimicrobial drugs.
Collapse
Affiliation(s)
- Jingwen Xue
- School of Medicine, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Yinxue Fu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Huang Li
- School of Medicine, Shanghai University, Shanghai, People's Republic of China
| | - Ting Zhang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Wei Cong
- School of Medicine, Shanghai University, Shanghai, People's Republic of China
| | - Honggang Hu
- School of Medicine, Shanghai University, Shanghai, People's Republic of China
| | - Zhiyuan Lu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Fang Yan
- School of Medicine, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Yulei Li
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| |
Collapse
|
9
|
Pattelli ON, Valdivia EM, Beyersdorf MS, Regan CS, Rivas M, Hebert KA, Merajver SD, Cierpicki T, Mapp AK. A Lipopeptidomimetic of Transcriptional Activation Domains Selectively Disrupts the Coactivator Med25 Protein-Protein Interactions. Angew Chem Int Ed Engl 2024; 63:e202400781. [PMID: 38527936 PMCID: PMC11134611 DOI: 10.1002/anie.202400781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
Short amphipathic peptides are capable of binding to transcriptional coactivators, often targeting the same binding surfaces as native transcriptional activation domains. However, they do so with modest affinity and generally poor selectivity, limiting their utility as synthetic modulators. Here we show that incorporation of a medium-chain, branched fatty acid to the N-terminus of one such heptameric lipopeptidomimetic (LPPM-8) increases the affinity for the coactivator Med25 >20-fold (Ki >100 μM to 4 μM), rendering it an effective inhibitor of Med25 protein-protein interactions (PPIs). The lipid structure, the peptide sequence, and the C-terminal functionalization of the lipopeptidomimetic each influence the structural propensity of LPPM-8 and its effectiveness as an inhibitor. LPPM-8 engages Med25 through interaction with the H2 face of its activator interaction domain and in doing so stabilizes full-length protein in the cellular proteome. Further, genes regulated by Med25-activator PPIs are inhibited in a cell model of triple-negative breast cancer. Thus, LPPM-8 is a useful tool for studying Med25 and mediator complex biology and the results indicate that lipopeptidomimetics may be a robust source of inhibitors for activator-coactivator complexes.
Collapse
Affiliation(s)
- Olivia N. Pattelli
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Estefanía Martínez Valdivia
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Matthew S. Beyersdorf
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Clint S. Regan
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Mónica Rivas
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | | | - Sofia D. Merajver
- Department of Internal Medicine, Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
| | - Tomasz Cierpicki
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
| | - Anna K. Mapp
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109 USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 USA
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
10
|
Mi T, Nguyen D, Gao Z, Burgess K. Bioinformatics leading to conveniently accessible, helix enforcing, bicyclic ASX motif mimics (BAMMs). Nat Commun 2024; 15:4217. [PMID: 38760359 PMCID: PMC11101637 DOI: 10.1038/s41467-024-48323-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Helix mimicry provides probes to perturb protein-protein interactions (PPIs). Helical conformations can be stabilized by joining side chains of non-terminal residues (stapling) or via capping fragments. Nature exclusively uses capping, but synthetic helical mimics are heavily biased towards stapling. This study comprises: (i) creation of a searchable database of unique helical N-caps (ASX motifs, a protein structural motif with two intramolecular hydrogen-bonds between aspartic acid/asparagine and following residues); (ii) testing trends observed in this database using linear peptides comprising only canonical L-amino acids; and, (iii) novel synthetic N-caps for helical interface mimicry. Here we show many natural ASX motifs comprise hydrophobic triangles, validate their effect in linear peptides, and further develop a biomimetic of them, Bicyclic ASX Motif Mimics (BAMMs). BAMMs are powerful helix inducing motifs. They are synthetically accessible, and potentially useful to a broad section of the community studying disruption of PPIs using secondary structure mimics.
Collapse
Affiliation(s)
- Tianxiong Mi
- Department of Chemistry, Texas A & M University, College Station, TX, 77842, USA
| | - Duyen Nguyen
- Department of Chemistry, Texas A & M University, College Station, TX, 77842, USA
| | - Zhe Gao
- Department of Chemistry, Texas A & M University, College Station, TX, 77842, USA
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, College Station, TX, 77842, USA.
| |
Collapse
|
11
|
Young R, Huang T, Luo Z, Tan YS, Kaur A, Lau YH. Development of stapled NONO-associated peptides reveals unexpected cell permeability and nuclear localisation. J Pept Sci 2024; 30:e3562. [PMID: 38148630 DOI: 10.1002/psc.3562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/28/2023]
Abstract
The non-POU domain-containing octamer-binding protein (NONO) is a nucleic acid-binding protein with diverse functions that has been identified as a potential cancer target in cell biology studies. Little is known about structural motifs that mediate binding to NONO apart from its ability to form homodimers, as well as heterodimers and oligomers with related homologues. We report a stapling approach to macrocyclise helical peptides derived from the insulin-like growth factor binding protein (IGFBP-3) that NONO interacts with, and also from the dimerisation domain of NONO itself. Using a range of chemistries including Pd-catalysed cross-coupling, cysteine arylation and cysteine alkylation, we successfully improved the helicity and observed modest peptide binding to the NONO dimer, although binding could not be saturated at micromolar concentrations. Unexpectedly, we observed cell permeability and preferential nuclear localisation of various dye-labelled peptides in live confocal microscopy, indicating the potential for developing peptide-based tools to study NONO in a cellular context.
Collapse
Affiliation(s)
- Reginald Young
- School of Chemistry, The University of Sydney, Camperdown, Australia
| | - Tiancheng Huang
- School of Chemistry, The University of Sydney, Camperdown, Australia
| | - Zijie Luo
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Matrix, Singapore
| | - Amandeep Kaur
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Melbourne, Australia
| | - Yu Heng Lau
- School of Chemistry, The University of Sydney, Camperdown, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, Australia
| |
Collapse
|
12
|
Mi T, Gao Z, Mituta Z, Burgess K. Dual-Capped Helical Interface Mimics. J Am Chem Soc 2024; 146:10331-10341. [PMID: 38573124 PMCID: PMC11027154 DOI: 10.1021/jacs.3c11717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 04/05/2024]
Abstract
Disruption of protein-protein interactions is medicinally important. Interface helices may be mimicked in helical probes featuring enhanced rigidities, binding to protein targets, stabilities in serum, and cell uptake. This form of mimicry is dominated by stapling between side chains of helical residues: there has been less progress on helical N-caps, and there were no generalizable C-caps. Conversely, in natural proteins, helicities are stabilized and terminated by C- and N-caps but not staples. Bicyclic caps previously introduced by us enable interface helical mimicry featuring rigid synthetic caps at both termini in this work. An unambiguously helical dual-capped system proved to be conformationally stable, binding cyclins A and E, and showed impressive cellular uptake. In addition, the dual-capped mimic was completely resistant to proteolysis in serum over an extended period when compared with "gold standard" hydrocarbon-stapled controls. Dual-capped peptidomimetics are a new, generalizable paradigm for helical interface probe design.
Collapse
Affiliation(s)
- Tianxiong Mi
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842, United States
| | - Zhe Gao
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842, United States
| | - Zeynep Mituta
- ZentriForce
Pharma Research GmbH, Carl-Friedrich-Gauss-Ring 5, 69124 Heidelberg, Germany
| | - Kevin Burgess
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842, United States
| |
Collapse
|
13
|
Kwok JG, Yuan Z, Arora PS. An Encodable Scaffold for Sequence-Specific Recognition of Duplex RNA. Angew Chem Int Ed Engl 2023; 62:e202308650. [PMID: 37548640 PMCID: PMC10528708 DOI: 10.1002/anie.202308650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/08/2023]
Abstract
RNA, unlike DNA, folds into a multitude of secondary and tertiary structures. This structural diversity has impeded the development of ligands that can sequence-specifically target this biomolecule. We sought to develop ligands for double-stranded RNA (dsRNA) segments, which are ubiquitous in RNA tertiary structure. The major groove of double-stranded DNA is sequence-specifically recognized by a range of dimeric helical transcription factors, including the basic leucine zippers (bZIP) and basic helix-loop-helix (bHLH) proteins; however, such simple structural motifs are not prevalent in RNA-binding proteins. We interrogated the high-resolution structures of DNA and RNA to identify requirements for a helix fork motif to occupy dsRNA major grooves akin to dsDNA. Our analysis suggested that the rigidity and angle of approach of dimeric helices in bZIP/bHLH motifs are not ideal for the binding of dsRNA major grooves. This investigation revealed that the replacement of the leucine zipper motifs in bHLH proteins with synthetic crosslinkers would allow recognition of dsRNA. We show that a model bHLH DNA-binding motif does not bind dsRNA but can be reengineered as an RNA ligand. Based on this hypothesis, we rationally designed a miniature synthetic crosslinked helix fork (CHF) as a generalizable proteomimetic scaffold for targeting dsRNA. We evaluated several CHF constructs against a set of RNA and DNA hairpins to probe the specificity of the designed construct. Our studies reveal a new class of proteomimetics as an encodable platform for sequence-specific recognition of dsRNA.
Collapse
Affiliation(s)
- Jonathan G. Kwok
- Department of Chemistry, New York University, 29 Washington Place, New York, NY10003
| | - Zhi Yuan
- Department of Chemistry, New York University, 29 Washington Place, New York, NY10003
| | - Paramjit S. Arora
- Department of Chemistry, New York University, 29 Washington Place, New York, NY10003
| |
Collapse
|
14
|
Victorio CG, Sawyer N. Folding-Assisted Peptide Disulfide Formation and Dimerization. ACS Chem Biol 2023; 18:1480-1486. [PMID: 37390465 DOI: 10.1021/acschembio.3c00268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Disulfide bonds form covalent bonds between distal regions of peptides and proteins to dramatically impact their folding, stability, and oligomerization. Given the prevalence of disulfide bonds in many natural products, considerable effort has been invested in site-selective disulfide bond formation approaches to control the folding of chemically synthesized peptides and proteins. Here, we show that the careful choice of thiol oxidation conditions can lead to monomeric or dimeric species from fully deprotected linear bisthiol peptides. Starting from a p53-derived peptide, we found that oxidation under aqueous (nondenaturing) conditions produces antiparallel dimers with enhanced α-helical character, while oxidation under denaturing conditions promotes formation of a nonhelical intramolecular disulfide species. Examination across peptide variants suggests that intramolecular disulfide formation is robust across diverse peptide sequences, while dimerization is sensitive to both the α-helical folding of the linear peptide and aromatic residues at the dimerization interface. All disulfide species are more resistant to protease degradation than the linear peptide but are easily reduced to restore the initial bisthiol peptide. Both disulfide formation approaches are compatible with α-helix-stabilizing cross-linkers. These results provide an approach for using disulfide bonds to control peptide folding and oligomerization to better understand how folding influences interactions with diverse molecular targets.
Collapse
Affiliation(s)
- Clara G Victorio
- Department of Chemistry, Fordham University, 441 E. Fordham Rd., Bronx, New York 10458, United States
| | - Nicholas Sawyer
- Department of Chemistry, Fordham University, 441 E. Fordham Rd., Bronx, New York 10458, United States
| |
Collapse
|
15
|
Duez Q, Hoyas S, Josse T, Cornil J, Gerbaux P, De Winter J. Gas-phase structure of polymer ions: Tying together theoretical approaches and ion mobility spectrometry. MASS SPECTROMETRY REVIEWS 2023; 42:1129-1151. [PMID: 34747528 DOI: 10.1002/mas.21745] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 06/07/2023]
Abstract
An increasing number of studies take advantage of ion mobility spectrometry (IMS) coupled to mass spectrometry (IMS-MS) to investigate the spatial structure of gaseous ions. Synthetic polymers occupy a unique place in the field of IMS-MS. Indeed, due to their intrinsic dispersity, they offer a broad range of homologous ions with different lengths. To help rationalize experimental data, various theoretical approaches have been described. First, the study of trend lines is proposed to derive physicochemical and structural parameters. However, the evaluation of data fitting reflects the overall behavior of the ions without reflecting specific information on their conformation. Atomistic simulations constitute another approach that provide accurate information about the ion shape. The overall scope of this review is dedicated to the synergy between IMS-MS and theoretical approaches, including computational chemistry, demonstrating the essential role they play to fully understand/interpret IMS-MS data.
Collapse
Affiliation(s)
- Quentin Duez
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
| | - Sébastien Hoyas
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
| | | | - Jérôme Cornil
- Laboratory for Chemistry of Novel Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
| | - Pascal Gerbaux
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
| | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory, Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons, UMONS, Mons, Belgium
| |
Collapse
|
16
|
Mi T, Nguyen D, Burgess K. Bicyclic Schellman Loop Mimics (BSMs): Rigid Synthetic C-Caps for Enforcing Peptide Helicity. ACS CENTRAL SCIENCE 2023; 9:300-306. [PMID: 36844493 PMCID: PMC9951308 DOI: 10.1021/acscentsci.2c01265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 06/18/2023]
Abstract
Macrocyclic peptides are the prevalent way to mimic interface helices for disruption of protein interactions, but current strategies to do this via synthetic C-cap mimics are underdeveloped and suboptimal. Bioinformatic studies described here were undertaken to better understand Schellman loops, the most common C-caps in proteins, to design superior synthetic mimics. An algorithm (Schellman Loop Finder) was developed, and data mining with this led to the discovery that these secondary structures are often stabilized by combinations of three hydrophobic side chains, most frequently from Leu, to form hydrophobic triangles. That insight facilitated design of synthetic mimics, bicyclic Schellman loop mimics (BSMs), where the hydrophobic triumvirate was replaced by 1,3,5-trimethylbenzene. We demonstrate that BSMs can be made quickly and efficiently, and are more rigid and helix-inducing than the best current C-cap mimics, which are rare and all monocycles.
Collapse
|
17
|
Design and Synthesis of Novel Helix Mimetics Based on the Covalent H-Bond Replacement and Amide Surrogate. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020780. [PMID: 36677838 PMCID: PMC9863496 DOI: 10.3390/molecules28020780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
A novel hydrogen bond surrogate-based (HBS) α-helix mimetic was designed by the combination of covalent H-bond replacement and the use of an ether linkage to substitute an amide bond within a short peptide sequence. The new helix template could be placed in position other than the N-terminus of a short peptide, and the CD studies demonstrate that the template adopts stable conformations in aqueous buffer at exceptionally high temperatures.
Collapse
|
18
|
Madheshiya PK, Shukla E, Singh J, Bawaria S, Ansari MY, Chauhan R. Insights into the role of Nup62 and Nup93 in assembling cytoplasmic ring and central transport channel of the nuclear pore complex. Mol Biol Cell 2022; 33:ar139. [PMID: 36222862 PMCID: PMC9727814 DOI: 10.1091/mbc.e22-01-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The nuclear pore complex (NPC) is a highly modular assembly of 34 distinct nucleoporins (Nups) to form a versatile transport channel between the nucleus and the cytoplasm. Among them, Nup62 is known as an essential component for nuclear transport, Nup93 for proper nuclear envelope assembly. These Nups constitute various NPC subcomplexes such as the central transport channel (CTC), the cytoplasmic ring (CR), and the inner ring (IR). However, how they play their roles in NPC assembly and transport activity is not clear. Here we delineated the interacting regions and conducted biochemical reconstitution and structural characterization of the mammalian CR complex to reveal its intrinsic dynamic behavior and a distinct "4"-shaped architecture resembling the CTC complex. Our in vitro reconstitution data demonstrate that the Nup62 coiled-coil domain is critical to form both Nup62322-525 •Nup88517-742 and Nup62322-525•Nup88517-742•Nup214693-926 heterotrimers and both can bind to Nup931-150. We therefore propose that Nup93 acts as a "sensor" to bind to Nup62 shared heterotrimers including the Nup62•Nup54 heterotrimer of the CTC, which was not shown previously to be an interacting partner. Altogether, our biochemical study suggests that Nup62 via its coiled-coil domain is central to form compositionally distinct yet structurally similar heterotrimers and Nup93 binds these diverse heterotrimers nonselectively.
Collapse
Affiliation(s)
| | - Ekta Shukla
- National Centre for Cell Science, Pune 411007, Maharashtra, India
| | - Jyotsana Singh
- National Centre for Cell Science, Pune 411007, Maharashtra, India
| | | | | | - Radha Chauhan
- National Centre for Cell Science, Pune 411007, Maharashtra, India,*Address correspondence to: Radha Chauhan ()
| |
Collapse
|
19
|
Whisenant J, Burgess K. Synthetic helical peptide capping strategies. Chem Soc Rev 2022; 51:5795-5804. [PMID: 35786712 DOI: 10.1039/d1cs01175h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Relatively small mimics of interface secondary structures can be used to disrupt protein-protein interactions (PPIs). This strategy is valuable because many PPIs are pivotal in cell biology and contemporary medicinal chemistry. Small peptides tend to have random coil conformations in solution, so the entropy costs are high for them to order into states binding protein receptors. Consequently, peptides constrained in conformations resembling interface secondary structures are favored for enhanced affinities to PPI components. Helices are commonly found at PPI interfaces. The two general strategies that have emerged for imposing helical constraints in probes, capping and stapling, are often confused because both involve formation of macrocyclic rings. This review considers parameters that distinguish capping from stapling. Capping motifs terminate helices and project the adjacent peptide units in non-helical orientations, but stapling enforces helical motifs in ways that enable adjacent peptide fragments to extend helices. There is no evidence that stapling is more effective than capping for helix mimicry, but stapling is used more frequently. This imbalance may be because no strategies have emerged for synthetic C-capping with compact unit; if convenient and effective C-capping strategies were available then capping strategies should be more widely used.
Collapse
Affiliation(s)
- Jonathan Whisenant
- Department of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842, USA.
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842, USA.
| |
Collapse
|
20
|
Su Z, Liu C, Cong W, He S, Su L, Hu H. Design, Synthesis, and Antitumor Activity Study of All-Hydrocarbon-Stapled B1-Leu Peptides. Front Chem 2022; 10:840131. [PMID: 35464194 PMCID: PMC9021566 DOI: 10.3389/fchem.2022.840131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
B1-Leu peptide is a structural optimization compound derived from the lysine- and phenylalanine-rich antimicrobial peptide Cathelicidin-BF. It has shown promising antibacterial and antitumor biological activity. However, linear peptides are not the best choice for novel drug development due to their poor pharmacokinetic properties. In this study, various all-hydrocarbon stapled B1-Leu derivatives were designed and synthesized. Their secondary structure, protease stability, and antitumor and hemolytic activities were also investigated to evaluate their clinical value for cancer therapy. Among them, B1-L-3 and B1-L-6 showed both damaging the tumor cell membrane stability and antitumor activity, showing that they are promising lead compounds for the development of novel cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
21
|
Tzotzos S. Stapled peptides as potential inhibitors of SARS‐CoV‐2 binding to the hACE2 receptor. J Pept Sci 2022; 28:e3409. [PMID: 35165970 PMCID: PMC9111031 DOI: 10.1002/psc.3409] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 12/03/2022]
Abstract
Stapled peptides are synthetic peptidomimetics of bioactive sites in folded proteins which carry chemical links, introduced during peptide synthesis, designed to retain the secondary structure in the native protein molecule. Stapled peptides have been investigated as potential modulators of protein–protein interactions for over two decades. The potential use of stapled peptides as inhibitors of viral entry, and therefore as antiviral therapeutics, has been established for several important viruses causing disease in humans, such as the human immunodeficiency virus type 1 (HIV‐1), respiratory syncytial virus (RSV), and Middle East Respiratory Syndrome (MERS) coronavirus. Several independent research initiatives have investigated the inhibitory effect of stapled peptides on binding of the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), the causative agent of COVID‐19, to its receptor, angiotensin‐converting‐enzyme 2 (ACE2). These stapled peptides, which mimic Helix 1 of the human ACE2 receptor, have demonstrated mixed ability to prevent infection with SARS‐CoV‐2 in cell‐based studies.
Collapse
|
22
|
Luong HX, Bui HTP, Tung TT. Application of the All-Hydrocarbon Stapling Technique in the Design of Membrane-Active Peptides. J Med Chem 2022; 65:3026-3045. [PMID: 35112864 DOI: 10.1021/acs.jmedchem.1c01744] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The threats of drug resistance and new emerging pathogens have led to an urgent need to develop alternative treatment therapies. Recently, considerable research efforts have focused on membrane-active peptides (MAPs), a category of peptides in drug discovery with antimicrobial, anticancer, and cell penetration activities that have demonstrated their potential to be multifunctional agents. Nonetheless, natural MAPs have encountered various disadvantages, which mainly include poor bioavailability, the lack of a secondary structure in short peptides, and high production costs for long peptide sequences. Hence, an "all-hydrocarbon stapling system" has been applied to these peptides and proven to effectively stabilize the helical conformations, improving proteolytic resistance and increasing both the potency and the cell permeability. In this review, we summarized and categorized the advances made using this powerful technique in the development of stapled MAPs. Furthermore, outstanding issues and suggestions for future design within each subcategory were thoroughly discussed.
Collapse
Affiliation(s)
- Huy Xuan Luong
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| | | | - Truong Thanh Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| |
Collapse
|
23
|
Chen K, Tang Y, Wu M, Wan XC, Zhang YN, Chen XX, Yu FQ, Cui ZH, Ma JM, Zhou Z, Fang GM. Head-to-Tail Cross-Linking to Generate Bicyclic Helical Peptides with Enhanced Helicity and Proteolytic Stability. Org Lett 2022; 24:53-57. [PMID: 34894695 DOI: 10.1021/acs.orglett.1c03629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report a new pattern of a bicyclic helical peptide constructed through head-to-tail cross-linking. The described bicyclic helical peptide has a head-to-tail cross-linking arm and a C-terminal i, i + 4 cross-linking arm. This scaffold will provide a promising scaffold for designing a proteolytically resistant helix-constrained peptide.
Collapse
Affiliation(s)
- Kai Chen
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Yang Tang
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, and Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai 200072, P. R. China
| | - Meng Wu
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Xiao-Cui Wan
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Yan-Ni Zhang
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Xiao-Xu Chen
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Fei-Qiang Yu
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Zhi-Hui Cui
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong 518055, P. R. China
| | - Jin-Ming Ma
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, P. R. China
| | - Ge-Min Fang
- School of Life Science, Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| |
Collapse
|
24
|
Yu Z, Tang H, Cong W, Gao F, Li H, Hu H, Wang X, He S. Hydrocarbon stapling modification of peptide alyteserin-2a: Discovery of novel stapled peptide antitumor agents. J Pept Sci 2022; 28:e3401. [PMID: 34989078 DOI: 10.1002/psc.3401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/24/2021] [Accepted: 12/24/2021] [Indexed: 11/10/2022]
Abstract
Alyteserin-2a (ILGKLLSTAAGLLSNL.NH2 ) is isolated from the skin exudates of midwife toad and has a wide range of biological applications. However, the use of alyteserin-2a as an antitumor agent is limited due to its structural flexibility. In this study, a series of stapled peptides were prepared through hydrocarbon stapling modification without destroying the key residues, and their chemical and biological properties were further evaluated for enhancing the application potential of alyteserin-2a in the field of antitumor drugs development. Among them, alyteserin-2a-Sp3 displayed significant improvement in helicity levels, protease resistance, and antitumor activity compared to that of the template peptide alyteserin-2a, indicating that alyteserin-2a-Sp3 had a potential to become a lead compound for the development of novel antitumor drugs. This study confirms the important effect of hydrocarbon stapling strategy on the secondary structure, hydrolase stability and biological activity of alyteserin-2a.
Collapse
Affiliation(s)
- Ziqiang Yu
- College of Sciences, Shanghai University, China
| | - Hua Tang
- Institute of Translational Medicine, Shanghai University, China
| | - Wei Cong
- Institute of Translational Medicine, Shanghai University, China
| | - Fei Gao
- Institute of Translational Medicine, Shanghai University, China
| | - Huaqiang Li
- Institute of Translational Medicine, Shanghai University, China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University, China
| | - Xiaoyan Wang
- Chongqing Health Center for Women and Children, Chongqing, China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University, China
| |
Collapse
|
25
|
Kim DI, Han SJ, Lim YB. Unique behaviour of α-helix in bending deformation. Chem Commun (Camb) 2022; 58:4368-4371. [DOI: 10.1039/d2cc00008c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Maximum degree of bending that can be tolerated by the rigid rod-like α-helix remains unknown; however, it should be very difficult or even impossible to make α-helices with varying degrees...
Collapse
|
26
|
A Rational Design of α-Helix-Shaped Peptides Employing the Hydrogen-Bond Surrogate Approach: A Modulation Strategy for Ras-RasGRF1 Interaction in Neuropsychiatric Disorders. Pharmaceuticals (Basel) 2021; 14:ph14111099. [PMID: 34832880 PMCID: PMC8623491 DOI: 10.3390/ph14111099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/09/2023] Open
Abstract
In the last two decades, abnormal Ras (rat sarcoma protein)-ERK (extracellular signal-regulated kinase) signalling in the brain has been involved in a variety of neuropsychiatric disorders, including drug addiction, certain forms of intellectual disability, and autism spectrum disorder. Modulation of membrane-receptor-mediated Ras activation has been proposed as a potential target mechanism to attenuate ERK signalling in the brain. Previously, we showed that a cell penetrating peptide, RB3, was able to inhibit downstream signalling by preventing RasGRF1 (Ras guanine nucleotide-releasing factor 1), a neuronal specific GDP/GTP exchange factor, to bind Ras proteins, both in brain slices and in vivo, with an IC50 value in the micromolar range. The aim of this work was to mutate and improve this peptide through computer-aided techniques to increase its inhibitory activity against RasGRF1. The designed peptides were built based on the RB3 peptide structure corresponding to the α-helix of RasGRF1 responsible for Ras binding. For this purpose, the hydrogen-bond surrogate (HBS) approach was exploited to maintain the helical conformation of the designed peptides. Finally, residue scanning, MD simulations, and MM-GBSA calculations were used to identify 18 most promising α-helix-shaped peptides that will be assayed to check their potential activity against Ras-RasGRF1 and prevent downstream molecular events implicated in brain disorders.
Collapse
|
27
|
Zheng M, Lupoli TJ. Modulation of a Mycobacterial ADP-Ribosyltransferase to Augment Rifamycin Antibiotic Resistance. ACS Infect Dis 2021; 7:2604-2611. [PMID: 34355905 DOI: 10.1021/acsinfecdis.1c00297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The rifamycins are broad-spectrum antibiotics that are primarily utilized to treat infections caused by mycobacteria, including tuberculosis. Interestingly, various species of bacteria are known to contain an enzyme called Arr that catalyzes ADP-ribosylation of rifamycin antibiotics as a mechanism of resistance. Here, we study Arr modulation in relevant Gram-positive and -negative species. We show that a C-terminal truncation of Arr (ArrC), encoded in the genome of Mycobacterium smegmatis, activates Arr-mediated rifamycin modification. Through structural comparisons of mycobacterial Arr and human poly(ADP-ribose) polymerases (PARPs), we identify a known small molecule PARP inhibitor that can act as an adjuvant to sensitize M. smegmatis to the rifamycin antibiotic rifampin via inhibition of Arr, even in the presence of ArrC. Finally, we demonstrate that this rifampin/adjuvant combination treatment is effective at inhibiting growth of the multidrug-resistant (MDR) nontuberculosis pathogen Mycobacterium abscessus, which has become a growing cause of human infections in the clinic.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Tania J. Lupoli
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
28
|
Zheng M, Wang R, Chen S, Zou Y, Yan L, Zhao L, Li X. Design, Synthesis and Antifungal Activity of Stapled Aurein1.2 Peptides. Antibiotics (Basel) 2021; 10:956. [PMID: 34439006 PMCID: PMC8389037 DOI: 10.3390/antibiotics10080956] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/18/2022] Open
Abstract
Aurein1.2 is a 13-residue antimicrobial peptide secreted by the Australian tree frog Litoria aurea. In order to improve its stabilities, the helical contents and corresponding biological activities of Aurein1.2 (a series of stapled analogues) were synthesized, and their potential antifungal activities were evaluated. Not surprisingly, the stapled Aurein1.2 peptides showed higher proteolytic stability and helicity than the linear counterpart. The minimum inhibitory concentration (MIC) of ten stapled peptides against six strains of common pathogenic fungi was determined by the microscale broth dilution method recommended by CLSI. Of them, Sau-1, Sau-2, Sau-5, and Sau-9 exhibited better inhibitory effects on the fungi than the linear peptide. These stapled Aurein1.2 peptides may serve as the leading compounds for further optimization and antifungal therapy.
Collapse
Affiliation(s)
- Mengjun Zheng
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China;
- School of Pharmacy, Naval Medical University, Shanghai 200433, China; (R.W.); (Y.Z.)
| | - Ruina Wang
- School of Pharmacy, Naval Medical University, Shanghai 200433, China; (R.W.); (Y.Z.)
| | - Si Chen
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Yan Zou
- School of Pharmacy, Naval Medical University, Shanghai 200433, China; (R.W.); (Y.Z.)
| | - Lan Yan
- School of Pharmacy, Naval Medical University, Shanghai 200433, China; (R.W.); (Y.Z.)
| | - Linjing Zhao
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China;
| | - Xiang Li
- School of Pharmacy, Naval Medical University, Shanghai 200433, China; (R.W.); (Y.Z.)
| |
Collapse
|
29
|
Jedhe GS, Arora PS. Hydrogen bond surrogate helices as minimal mimics of protein α-helices. Methods Enzymol 2021; 656:1-25. [PMID: 34325784 DOI: 10.1016/bs.mie.2021.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Examination of complexes of proteins with biomolecular ligands reveals that proteins tend to interact with partners via folded sub-domains, in which the backbone possesses secondary structure. α-Helices comprising the largest class of protein secondary structures, play fundamental roles in a multitude of highly specific protein-protein and protein-nucleic acid interactions. We have demonstrated a unique strategy for stabilization of the α-helical conformation that involves replacement of one of the main chain i and i+4 hydrogen bonds in the target α-helix with a covalent bond. We termed this synthetic strategy a hydrogen bond surrogate (HBS) approach. Two salient features of this approach are: (1) the internal placement of the crosslink allows development of helices such that none of the solvent-exposed surfaces are blocked by the constraining element, i.e., all side chains of the constrained helices remain available for molecular recognition. (2) This approach can be deployed to constrain very short peptides (<10 amino acid residues) into highly stable α-helices. This chapter presents the biophysical basis for the development of the hydrogen bond surrogate approach, as well as methods for the synthesis and conformational analysis of the artificial helices.
Collapse
Affiliation(s)
- Ganesh S Jedhe
- Department of Chemistry, New York University, New York, NY, United States
| | - Paramjit S Arora
- Department of Chemistry, New York University, New York, NY, United States.
| |
Collapse
|
30
|
Ricardo MG, Vázquéz-Mena Y, Iglesias-Morales Y, Wessjohann LA, Rivera DG. On the scope of the double Ugi multicomponent stapling to produce helical peptides. Bioorg Chem 2021; 113:104987. [PMID: 34022444 DOI: 10.1016/j.bioorg.2021.104987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/19/2021] [Accepted: 05/08/2021] [Indexed: 11/15/2022]
Abstract
The stabilization of helical structures by peptide stapling approaches is now a mature technology capable to provide a variety of biomedical applications. Recently, it was shown that multicomponent macrocyclization is not only an effective way to introduce conformational constraints but it also allows to incorporate additional functionalities to the staple moiety in a one-pot process. This work investigates the scope of the double Ugi multicomponent stapling approach in its capacity to produce helical peptides from unstructured sequences. For this, three different stapling combinations were implemented and the CD spectra of the cyclic peptides were measured to determine the effect of the multicomponent macrocyclization on the resulting secondary structure. A new insight into some structural factors influencing the helicity type and content is provided, along with new prospects on the utilization of this methodology to diversify the molecular tethers linking the amino acid side chains.
Collapse
Affiliation(s)
- Manuel G Ricardo
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba; Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| | - Yadiel Vázquéz-Mena
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | - Yuleidys Iglesias-Morales
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany.
| | - Daniel G Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba; Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
31
|
Pal S, Prabhakaran EN. Trimodular Solution‐Phase Protocol for Rapid Large‐Scale Synthesis of Hydrogen Bond Surrogate‐Constrained α‐Helicomimics. European J Org Chem 2021. [DOI: 10.1002/ejoc.202001359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Sunit Pal
- Department of Organic Chemistry Indian Institute of Science Bangalore Karnataka 560012 India
| | - Erode N. Prabhakaran
- Department of Organic Chemistry Indian Institute of Science Bangalore Karnataka 560012 India
| |
Collapse
|
32
|
Besenius P, Zengerling L, Kemper B, Hellmich UA. Synthesis and Structural Stability of α-Helical Gold(I)-Metallopeptidesy. Synlett 2021. [DOI: 10.1055/a-1290-8412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractThe synthesis of hexa- and dodecapeptides functionalized with two Au(I)–phosphine complexes is reported. The high stability of the Au(I)–phosphine bond allowed orthogonal peptide-protecting-group chemistry, even when using hard Lewis acids like boron tribromide. This enabled the preparation of an Fmoc-protected lysine derivative carrying the Au(I) complex in a side chain, which was used in standard Fmoc-based solid-phase peptide synthesis protocols. Alanine and leucine repeats in the metallododecapeptide formed α-helical secondary structures in 2,2,2-trifluoroethanol–H2O and 1,1,1,3,3,3-hexafluoroisopropanol–H2O mixtures with high thermal stability, as shown by temperature-dependent CD spectroscopy studies.
Collapse
Affiliation(s)
- Pol Besenius
- Department of Chemistry, Johannes Gutenberg-University Mainz
| | | | - Benedict Kemper
- Department of Chemistry, Johannes Gutenberg-University Mainz
| | - Ute A. Hellmich
- Department of Chemistry, Johannes Gutenberg-University Mainz
- Centre for Biomolecular Magnetic Resonance (BMRZ), Goethe-University Frankfurt
| |
Collapse
|
33
|
Wang N, Xie G, Liu C, Cong W, He S, Li Y, Fan L, Hu HG. Design, Synthesis, and Antitumor Activities Study of Stapled A4K14-Citropin 1.1 Peptides. Front Chem 2020; 8:616147. [PMID: 33363118 PMCID: PMC7758422 DOI: 10.3389/fchem.2020.616147] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/11/2020] [Indexed: 02/05/2023] Open
Abstract
A4K14-citropin 1.1 is a structurally optimized derivative derived from amphibians' skin secreta peptide Citropin, which exhibits broad biological activities. However, the application of A4K14-citropin 1.1 as a cancer therapeutic is restricted by its structural flexibility. In this study, a series of all-hydrocarbon stapled peptides derivatives of A4K14-citropin 1.1 were designed and synthesized, and their chemical and biological characteristics were also investigated. Among them, A4K14-citropin 1.1-Sp1 and A4K14-citropin 1.1-Sp4 displayed improved helicity levels, greater protease stability, and increased antitumor activity compared with the original peptide, which establishes them as promising lead compounds for novel cancer therapeutics development. These results revealed the important influence of all-hydrocarbon stapling side chain on the secondary structure, hydrolase stability, and biological activity of A4K14-citropin 1.1.
Collapse
Affiliation(s)
- Nan Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Gang Xie
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Chao Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Wei Cong
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Li Fan
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hong-Gang Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
34
|
Pal S, Banerjee S, Prabhakaran EN. Helix-Coil Transition at a Glycine Following a Nascent α-Helix: A Synergetic Guidance Mechanism for Helix Growth. J Phys Chem A 2020; 124:7478-7490. [PMID: 32877193 DOI: 10.1021/acs.jpca.0c05489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A detailed understanding of forces guiding the rapid folding of a polypeptide from an apparently random coil state to an ordered α-helical structure following the rate-limiting preorganization of the initial three residue backbones into helical conformation is imperative to comprehending and regulating protein folding and for the rational design of biological mimetics. However, several details of this process are still unknown. First, although the helix-coil transition was proposed to originate at the residue level (J. Chem. Phys. 1959, 31, 526-535; J. Chem. Phys. 1961, 34, 1963-1974), all helix-folding studies have only established it between time-averaged bulk states of a long-lived helix and several transiently populated random coils, along the whole helix model sequence. Second, the predominant thermodynamic forces driving either this two-state transition or the faster helix growth following helix nucleation are still unclear. Third, the conformational space of the random coil state is not well-defined unlike its corresponding α-helix. Here we investigate the restrictions placed on the conformational space of a Gly residue backbone, as a result of it immediately succeeding a nascent α-helical turn. Analyses of the temperature-dependent 1D-, 2D-NMR, FT-IR, and CD spectra and GROMACS MD simulation trajectory of a Gly residue backbone following a model α-helical turn, which is artificially rigidified by a covalent hydrogen bond surrogate, reveal that: (i) the α-helical turn guides the ϕ torsion of the Gly exclusively into either a predominantly populated entropically favored α-helical (α-ϕ) state or a scarcely populated random coil (RC-ϕ) state; (ii) the α-ϕ state of Gly in turn favors the stability of the preceding α-helical turn, while the RC-ϕ state disrupts it, revealing an entropy-driven synergetic guidance for helix growth in the residue following helix nucleation. The applicability of a current synergetic guidance mechanism to explain rapid helix growth in folded and unfolded states of proteins and helical peptides is discussed.
Collapse
Affiliation(s)
- Sunit Pal
- Department of Chemistry, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Shreya Banerjee
- Department of Chemistry, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Erode N Prabhakaran
- Department of Chemistry, Indian Institute of Science, Bangalore, Karnataka-560012, India
| |
Collapse
|
35
|
Outlaw VK, Kreitler DF, Stelitano D, Porotto M, Moscona A, Gellman SH. Effects of Single α-to-β Residue Replacements on Recognition of an Extended Segment in a Viral Fusion Protein. ACS Infect Dis 2020; 6:2017-2022. [PMID: 32692914 PMCID: PMC8019249 DOI: 10.1021/acsinfecdis.0c00385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Partial replacement of α-amino acid residues with β-amino acid residues has been established as a strategy for preserving target-engagement by helix-forming polypeptides while altering other properties. The impact of β-residue incorporation within polypeptides that adopt less regular conformations, however, has received less attention. The C-terminal heptad repeat (HRC) domains of fusion glycoproteins from pathogenic paramyxoviruses contain a segment that must adopt an extended conformation in order to coassemble with the N-terminal heptad repeat (HRN) domain in the postfusion state and drive a merger of the viral envelope with a target cell membrane. Here, we examine the impact of single α-to-β substitutions within this extended N-terminal segment of an engineered HRC peptide designated VIQKI. Stabilities of hexameric coassemblies formed with the native human parainfluenza virus 3 (HPIV3) HRN have been evaluated, the structures of five coassemblies have been determined, and antiviral efficacies have been measured. Many sites within the extended segment show functional tolerance of α-to-β substitution. These results offer a basis for future development of paramyxovirus infection inhibitors with novel biological activity profiles, possibly including resistance to proteolysis.
Collapse
Affiliation(s)
- Victor K. Outlaw
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, 53706, United States
| | - Dale F. Kreitler
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, 53706, United States
| | - Debora Stelitano
- Department of Pediatrics, Columbia University Medical Center, New York, New York, 10032, United States
- Center for Host–Pathogen Interaction, Columbia University Medical Center, New York, New York, 10032, United States
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Matteo Porotto
- Department of Pediatrics, Columbia University Medical Center, New York, New York, 10032, United States
- Center for Host–Pathogen Interaction, Columbia University Medical Center, New York, New York, 10032, United States
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Anne Moscona
- Department of Pediatrics, Columbia University Medical Center, New York, New York, 10032, United States
- Center for Host–Pathogen Interaction, Columbia University Medical Center, New York, New York, 10032, United States
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, New York, 10032, United States
- Department of Physiology & Cellular Biophysics, Columbia University Medical Center, New York, New York, 10032, United States
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, 53706, United States
| |
Collapse
|
36
|
Wang N, Li J, Song H, Liu C, Hu H, Liao H, Cong W. Synthesis and anti-osteoporosis activity of novel Teriparatide glycosylation derivatives. RSC Adv 2020; 10:25730-25735. [PMID: 35518599 PMCID: PMC9055339 DOI: 10.1039/d0ra05136e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is a metabolic bone disease that is characterized by low bone mass and micro-architectural deterioration of bones. The mechanism underlying this disease implicates an imbalance between bone resorption and bone remodeling. In 2002, the US Food and Drug Administration (FDA) approved Teriparatide for the treatment of osteoporosis, and so far, this compound is the only permitted osteoanabolic. However, as a structurally flexible linear peptide, this drug may be further optimized. In this study, we develop a series of novel N-acetyl glucosamine glycosylation derivatives of Teriparatide and examine their characteristics. Of the analyzed compounds, PTHG-9 exhibits enhanced helicity, greater protease stability, and increased osteoblast differentiation promoting ability compared with the original Teriparatide. Accordingly, PTHG-9 is suggested as a therapeutic candidate for postmenopausal osteoporosis (PMOP) and other related diseases. The successful development of an enhanced osteoporosis drug proves that the method proposed herein can be used to effectively enhance the chemical and biological properties of linear peptides with various biological functions.
Collapse
Affiliation(s)
- Nan Wang
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Jingyang Li
- Department of Pediatric Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hui Song
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Chao Liu
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Hongli Liao
- School of Pharmacy, Chengdu Medical College Chengdu China
| | - Wei Cong
- Institute of Translational Medicine, Shanghai University Shanghai China
| |
Collapse
|
37
|
Pal S, Banerjee S, Kumar A, Prabhakaran EN. H-Bond Surrogate-Stabilized Shortest Single-Turn α-Helices: sp 2 Constraints and Residue Preferences for the Highest α-Helicities. ACS OMEGA 2020; 5:13902-13912. [PMID: 32566857 PMCID: PMC7301546 DOI: 10.1021/acsomega.0c01277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/20/2020] [Indexed: 05/08/2023]
Abstract
Short α-helical sequences of proteins fail to maintain their native conformation when taken out of their protein context. Several covalent constraints have been designed, including the covalent H-bond surrogate (HBS)-where a peptide backbone i + 4 → i H-bond is replaced by a covalent surrogate-to nucleate α-helix in short sequences (>7 < 15 amino acids). But constraining the shortest sequences (four amino acids) into a single α-helical turn is still a significant challenge. Here, we introduce an HBS model that can be placed in unstructured tetrapeptides without excising any of its residues, and that biases them predominantly into remarkably stable single α-helical turns in varying solvents, pH values, and temperatures. Circular dichroism (CD), Fourier transform infrared (FT-IR) absorption, one-dimensional (1D)-NMR, two-dimensional (2D)-NMR spectral and computational analyses of the HBS-constrained tetrapeptide analogues reveal that (a) the number of sp2 atoms in the HBS-constrained backbone influences their predominance and rigidity in the α-helical conformation; and (b) residue preferences at the unnatural HBS-constrained positions influence their α-helicities, with Moc[GFA]G-OMe (1a) showing the highest known α-helicity (θn→π*MRE ∼-25.3 × 103 deg cm2 dmol-1 at 228 nm) for a single α-helical turn. Current findings benefit chemical biological applications desiring predictable access to single α-helical turns in tetrapeptides.
Collapse
|
38
|
Jerath G, Goyal R, Trivedi V, Santhoshkumar TR, Ramakrishnan V. Conformationally constrained peptides for drug delivery. J Pept Sci 2020; 26:e3244. [PMID: 32128940 DOI: 10.1002/psc.3244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/26/2022]
Abstract
Peptides have shown great potential in acting as template for developing versatile carrier platforms in nanomedicine, aimed at selective delivery of drugs to only pathological tissues saving its normal neighbors. Cell-penetrating peptides (CPPs) are short oligomeric peptides capable of translocating across the cell membrane while simultaneously employing multiple mechanisms of entry. Most CPPs exist as disordered structures in solution and may adopt a helical conformation on interaction with cell membrane, vital to their penetrative capability. Herein, we report a series of cationic helical amphipathic peptides (CHAPs), which are topologically constrained to be helical. The peptides were tested against cervical and breast cancer cells for their cell penetration and drug delivery potential. The cellular uptake of CHAP peptides is independent of temperature and energy availability. The activity of the peptides is biocompatible in bovine serum. CHAPs delivered functional methotrexate (MTX) inside the cell as CHAP-MTX conjugates. CHAP-MTX conjugates were more toxic to cancer cells than MTX alone. However, the CHAP-MTX conjugates were less toxic to HEK-293 cells compared with the cancer cells suggesting higher affinity towards cancer cells.
Collapse
Affiliation(s)
- Gaurav Jerath
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Ruchika Goyal
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | | | - Vibin Ramakrishnan
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
39
|
Dai Y, Yue N, Gong J, Liu C, Li Q, Zhou J, Huang W, Qian H. Development of cell-permeable peptide-based PROTACs targeting estrogen receptor α. Eur J Med Chem 2019; 187:111967. [PMID: 31865016 DOI: 10.1016/j.ejmech.2019.111967] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 01/27/2023]
Abstract
Proteolysis-targeting chimera (PROTAC) could selectively degrade target protein and may become a promising strategy for treating estrogen receptor α (ERα) positive breast cancers. Here, we designed penetrated peptide-based PROTACs by constructing an N-terminal lactam cyclic to improve proteolytic stability and cell penetration. We used a lactam cyclic peptide as ERα binding ligand, 6-aminocaproic acid as a linker, and a hydroxylated pentapeptide structure for recruiting E3 ligase to obtain heterobifunctional compounds. The resulting optimized compound I-6 selectively recruited ERα to the E3 ligase complex for promoting the degradation of ERα. Compound I-6 possessed strong effect on MCF-7 cell toxicity (IC50 ∼9.7 μM) and significantly enhanced activities in inducing ERα degradation. Meanwhile, I-6 performed much stronger potency in inhibition of tumors growth than tamoxifen. This work is a successful template to construct PROTACs based on cell-permeable peptides, which could extend the chemical space of PROTACs.
Collapse
Affiliation(s)
- Yuxuan Dai
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, 710021, PR China
| | - Na Yue
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Junni Gong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Chunxia Liu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Qifei Li
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Jiaqi Zhou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
40
|
Botta J, Bibic L, Killoran P, McCormick PJ, Howell LA. Design and development of stapled transmembrane peptides that disrupt the activity of G-protein-coupled receptor oligomers. J Biol Chem 2019; 294:16587-16603. [PMID: 31467080 PMCID: PMC6851324 DOI: 10.1074/jbc.ra119.009160] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/08/2019] [Indexed: 12/23/2022] Open
Abstract
Membrane proteins can associate into larger complexes. Examples include receptor tyrosine complexes, ion channels, transporters, and G protein–coupled receptors (GPCRs). For the latter, there is abundant evidence indicating that GPCRs assemble into complexes, through both homo- and heterodimerization. However, the tools for studying and disrupting these complexes, GPCR or otherwise, are limited. Here, we have developed stabilized interference peptides for this purpose. We have previously reported that tetrahydrocannabinol-mediated cognitive impairment arises from homo- or heterooligomerization between the GPCRs cannabinoid receptor type 1 (CB1R) and 5-hydroxytryptamine 2A (5-HT2AR) receptors. Here, to disrupt this interaction through targeting CB1–5-HT2A receptor heteromers in HEK293 cells and using an array of biochemical techniques, including calcium and cAMP measurements, bimolecular fluorescence complementation assays, and CD-based helicity assessments, we developed a NanoLuc binary technology (NanoBiT)-based reporter assay to screen a small library of aryl-carbon–stapled transmembrane-mimicking peptides produced by solid-phase peptide synthesis. We found that these stapling peptides have increased α-helicity and improved proteolytic resistance without any loss of disrupting activity in vitro, suggesting that this approach may also have utility in vivo. In summary, our results provide proof of concept for using NanoBiT to study membrane protein complexes and for stabilizing disrupting peptides to target such membrane complexes through hydrocarbon-mediated stapling. We propose that these peptides could be developed to target previously undruggable GPCR heteromers.
Collapse
Affiliation(s)
- Joaquín Botta
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.,School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Lucka Bibic
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Patrick Killoran
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Lesley A Howell
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
41
|
Dhaked HPS, Ray S, Battaje RR, Banerjee A, Panda D. Regulation ofStreptococcus pneumoniaeFtsZ assembly by divalent cations: paradoxical effects of Ca2+on the nucleation and bundling of FtsZ polymers. FEBS J 2019; 286:3629-3646. [DOI: 10.1111/febs.14928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/14/2019] [Accepted: 05/13/2019] [Indexed: 01/10/2023]
Affiliation(s)
| | - Shashikant Ray
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay India
- Department of Biotechnology Mahatma Gandhi Central University Motihari Bihar India
| | - Rachana Rao Battaje
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay India
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay India
| | - Dulal Panda
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay India
| |
Collapse
|
42
|
Guarracino DA, Riordan JA, Barreto GM, Oldfield AL, Kouba CM, Agrinsoni D. Macrocyclic Control in Helix Mimetics. Chem Rev 2019; 119:9915-9949. [DOI: 10.1021/acs.chemrev.8b00623] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Danielle A. Guarracino
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Jacob A. Riordan
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Gianna M. Barreto
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Alexis L. Oldfield
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Christopher M. Kouba
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Desiree Agrinsoni
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| |
Collapse
|
43
|
Design and structural characterisation of monomeric water-soluble α-helix and β-hairpin peptides: State-of-the-art. Arch Biochem Biophys 2019; 661:149-167. [DOI: 10.1016/j.abb.2018.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
|
44
|
Ricardo MG, Marrrero JF, Valdés O, Rivera DG, Wessjohann LA. A Peptide Backbone Stapling Strategy Enabled by the Multicomponent Incorporation of Amide N-Substituents. Chemistry 2018; 25:769-774. [PMID: 30412333 DOI: 10.1002/chem.201805318] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Indexed: 12/12/2022]
Abstract
The multicomponent backbone N-modification of peptides on solid-phase is presented as a powerful and general method to enable peptide stapling at the backbone instead of the side chains. This work shows that a variety of functionalized N-substituents suitable for backbone stapling can be readily introduced by means of on-resin Ugi multicomponent reactions conducted during solid-phase peptide synthesis. Diverse macrocyclization chemistries were implemented with such backbone N-substituents, including the ring-closing metathesis, lactamization, and thiol alkylation. The backbone N-modification method was also applied to the synthesis of α-helical peptides by linking N-substituents to the peptide N-terminus, thus featuring hydrogen-bond surrogate structures. Overall, the strategy proves useful for peptide backbone macrocyclization approaches that show promise in peptide drug discovery.
Collapse
Affiliation(s)
- Manuel G Ricardo
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle/Saale, Germany.,Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana, Cuba
| | - Javiel F Marrrero
- Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana, Cuba
| | - Oscar Valdés
- Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, 3460000, Chile
| | - Daniel G Rivera
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle/Saale, Germany.,Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana, Cuba
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle/Saale, Germany
| |
Collapse
|
45
|
Qin W, Xie M, Qin X, Fang Q, Yin F, Li Z. Recent advances in peptidomimetics antagonists targeting estrogen receptor α-coactivator interaction in cancer therapy. Bioorg Med Chem Lett 2018; 28:2827-2836. [DOI: 10.1016/j.bmcl.2018.05.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/25/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023]
|
46
|
Yang D, Qin W, Shi X, Zhu B, Xie M, Zhao H, Teng B, Wu Y, Zhao R, Yin F, Ren P, Liu L, Li Z. Stabilized β-Hairpin Peptide Inhibits Insulin Degrading Enzyme. J Med Chem 2018; 61:8174-8185. [DOI: 10.1021/acs.jmedchem.8b00418] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Dan Yang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
- Department of Science & Technology of Shandong Province, Jinan 250101, Shandong, China
| | - Weirong Qin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Xiaodong Shi
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Bili Zhu
- School of Medicine, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Mingsheng Xie
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Hui Zhao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Bin Teng
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yujie Wu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Rongtong Zhao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Peigen Ren
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Lizhong Liu
- School of Medicine, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| |
Collapse
|
47
|
Guo Y, Fu L, Fan X, Shi X. Stapled SC34EK fusion inhibitors with high potency against HIV-1 and improved protease resistance. CHINESE CHEM LETT 2018. [DOI: 10.1016/j.cclet.2018.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Qu Q, Pan M, Gao S, Zheng Q, Yu Y, Su J, Li X, Hu H. A Highly Efficient Synthesis of Polyubiquitin Chains. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800234. [PMID: 30027052 PMCID: PMC6051384 DOI: 10.1002/advs.201800234] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/17/2018] [Indexed: 06/08/2023]
Abstract
A robust, microwave-assisted, highly efficient, solid-phase peptide synthesis method for preparing isopeptide-linked 62-mer and 76-mer isoubiquitins and polyubiquitin is developed. The strategy avoids the use of costly resins and pseudoprolines, and the isopeptide-linked building blocks can be assembled with high initial purity within 1 day. All seven diubiquitins are successfully synthesized on a multi-milligram scale; a four-segment, three-ligation method is used to obtain a K33-/K11-linked mixed triubiquitin in excellent yield. Circular dichroism and crystallographic analyses are used to verify the structures of the well-folded, synthetic polyubiquitin chains. The facile synthetic strategy is expected to be generally applicable for the rapid synthesis of isopeptide-linked isoUbs and to pave the way for the study of longer polyubiquitin chains.
Collapse
Affiliation(s)
- Qian Qu
- School of PharmacySecond Military Medical University325 Guohe RoadShanghai200433China
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Man Pan
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Shuai Gao
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Qing‐Yun Zheng
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Yuan‐Yuan Yu
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Jia‐Can Su
- Changhai HospitalSecond Military Medical University168 Changhai RoadShanghai200433China
| | - Xiang Li
- School of PharmacySecond Military Medical University325 Guohe RoadShanghai200433China
| | - Hong‐Gang Hu
- School of PharmacySecond Military Medical University325 Guohe RoadShanghai200433China
| |
Collapse
|
49
|
Guo Y, Zhou PP, Zhang SY, Fan XW, Dou YW, Shi XL. Generation of a long-acting fusion inhibitor against HIV-1. MEDCHEMCOMM 2018; 9:1226-1231. [PMID: 30109011 DOI: 10.1039/c8md00124c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/29/2018] [Indexed: 11/21/2022]
Abstract
AIDS has evolved from a fatal infectious disease to a manageable chronic disease under the treatment of anti-AIDS medications. HIV fusion inhibitors with high activity, low side effects and strong selectivity are promising drugs against HIV. Only one fusion inhibitor is currently approved, thereby highly active long-acting fusion inhibitors need to be developed for long-term AIDS treatment. Here, we synthesised MT-SC22EK (a small HIV fusion inhibitor) derivatives containing 1-2 staples to improve its stability. Antiviral activity studies showed that MT-SC22EK-2 with two staples exhibited potent inhibitory activity against HIV-1 standard strains and Chinese epidemic strains, and at the same time, MT-SC22EK-2 presented strong anti-T20 resistance. Surprisingly, MT-SC22EK-2 possessed excellent protease stability with a half-life of 3665 min. MT-SC22EK-2 is a potential HIV fusion inhibitor considered as a long-acting anti-HIV drug candidate.
Collapse
Affiliation(s)
- Ye Guo
- School of Pharmacy , Baotou Medical College , Baotou 014060 , China
| | - Pan-Pan Zhou
- Comprehensive AIDS Research Center , School of Medicine , Tsinghua University , Beijing 100084 , China .
| | - Sen-Yan Zhang
- Comprehensive AIDS Research Center , School of Medicine , Tsinghua University , Beijing 100084 , China .
| | - Xiao-Wen Fan
- School of Pharmacy , Baotou Medical College , Baotou 014060 , China
| | - Yu-Wei Dou
- School of Pharmacy , Baotou Medical College , Baotou 014060 , China
| | - Xuan-Ling Shi
- Comprehensive AIDS Research Center , School of Medicine , Tsinghua University , Beijing 100084 , China .
| |
Collapse
|
50
|
Jiang Y, Deng Q, Zhao H, Xie M, Chen L, Yin F, Qin X, Zheng W, Zhao Y, Li Z. Development of Stabilized Peptide-Based PROTACs against Estrogen Receptor α. ACS Chem Biol 2018; 13:628-635. [PMID: 29271628 DOI: 10.1021/acschembio.7b00985] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peptide modulators targeting protein-protein interactions (PPIs) exhibit greater potential than small-molecule drugs in several important aspects including facile modification and relative large contact surface area. Stabilized peptides constructed by variable chemistry methods exhibit improved peptide stability and cell permeability compared to that of the linears. Herein, we designed a stabilized peptide-based proteolysis-targeting chimera (PROTAC) targeting estrogen receptor α (ERα) by tethering an N-terminal aspartic acid cross-linked stabilized peptide ERα modulator (TD-PERM) with a pentapeptide that binds the Von Hippel-Lindau (VHL) E3 ubiquitin ligase complex. The resulting heterobifunctional peptide (TD-PROTAC) selectively recruits ERα to the VHL E3 ligase complex, leading to the degradation of ERα in a proteasome-dependent manner. Compared with the control peptides, TD-PROTAC shows significantly enhanced activities in reducing the transcription of the ERα-downstream genes and inhibiting the proliferation of ERα-positive breast cancer cells. In addition, in vivo experiments indicate that TD-PROTAC leads to tumor regression in the MCF-7 mouse xenograft model. This work is a successful attempt to construct PROTACs based on cell-permeable stabilized peptides, which significantly broadens the chemical space of PROTACs and stabilized peptides.
Collapse
Affiliation(s)
- Yanhong Jiang
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Qiwen Deng
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Hui Zhao
- Division of Life Sciences, Clarivate Analytics, Beijing, 100190, China
| | - Mingsheng Xie
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Longjian Chen
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Feng Yin
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Xuan Qin
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Weihao Zheng
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Yongjuan Zhao
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| | - Zigang Li
- Key Lab of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
| |
Collapse
|